Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Nature ; 526(7573): 453-7, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26444240

RESUMO

Activation of oncogenes by mechanisms other than genetic aberrations such as mutations, translocations, or amplifications is largely undefined. Here we report a novel isoform of the anaplastic lymphoma kinase (ALK) that is expressed in ∼11% of melanomas and sporadically in other human cancer types, but not in normal tissues. The novel ALK transcript initiates from a de novo alternative transcription initiation (ATI) site in ALK intron 19, and was termed ALK(ATI). In ALK(ATI)-expressing tumours, the ATI site is enriched for H3K4me3 and RNA polymerase II, chromatin marks characteristic of active transcription initiation sites. ALK(ATI) is expressed from both ALK alleles, and no recurrent genetic aberrations are found at the ALK locus, indicating that the transcriptional activation is independent of genetic aberrations at the ALK locus. The ALK(ATI) transcript encodes three proteins with molecular weights of 61.1, 60.8 and 58.7 kilodaltons, consisting primarily of the intracellular tyrosine kinase domain. ALK(ATI) stimulates multiple oncogenic signalling pathways, drives growth-factor-independent cell proliferation in vitro, and promotes tumorigenesis in vivo in mouse models. ALK inhibitors can suppress the kinase activity of ALK(ATI), suggesting that patients with ALK(ATI)-expressing tumours may benefit from ALK inhibitors. Our findings suggest a novel mechanism of oncogene activation in cancer through de novo alternative transcription initiation.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Neoplasias/enzimologia , Neoplasias/genética , Receptores Proteína Tirosina Quinases/genética , Iniciação da Transcrição Genética , Alelos , Quinase do Linfoma Anaplásico , Animais , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Feminino , Células HEK293 , Histonas/química , Histonas/metabolismo , Humanos , Íntrons/genética , Isoenzimas/antagonistas & inibidores , Isoenzimas/biossíntese , Isoenzimas/química , Isoenzimas/genética , Lisina/metabolismo , Metilação , Camundongos , Dados de Sequência Molecular , Peso Molecular , Células NIH 3T3 , Neoplasias/tratamento farmacológico , Oncogenes/genética , Estrutura Terciária de Proteína/genética , RNA Polimerase II/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/genética , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/biossíntese , Receptores Proteína Tirosina Quinases/química , Transdução de Sinais
2.
Semin Cell Dev Biol ; 30: 75-85, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24735648

RESUMO

The transport of germ cells across the seminiferous epithelium is composed of a series of cellular events during the epithelial cycle essential to the completion of spermatogenesis. Without the timely transport of spermatids during spermiogenesis, spermatozoa that are transformed from step 19 spermatids in the rat testis fail to reach the luminal edge of the apical compartment and enter the tubule lumen at spermiation, thereby arriving the epididymis for further maturation. Step 19 spermatids and/or sperms that remain in the epithelium beyond stage VIII of the epithelial cycle will be removed by the Sertoli cell via phagocytosis to form phagosomes and be degraded by lysosomes, leading to subfertility and/or infertility. However, the biology of spermatid transport, in particular the final events that lead to spermiation remain elusive. Based on recent data in the field, we critically evaluate the biology of spermiation herein by focusing on the actin binding proteins (ABPs) that regulate the organization of actin microfilaments at the Sertoli-spermatid interface, which is crucial for spermatid transport during this event. The hypothesis we put forth herein also highlights some specific areas of research that can be pursued by investigators in the years to come.


Assuntos
Proteínas dos Microfilamentos/fisiologia , Espermátides/fisiologia , Actinas/fisiologia , Animais , Humanos , Masculino , Multimerização Proteica , Transdução de Sinais , Transporte Espermático
3.
Semin Cell Dev Biol ; 30: 65-74, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24727349

RESUMO

Non-receptor protein tyrosine kinases are cytoplasmic kinases that activate proteins by phosphorylating tyrosine residues, which in turn affect multiple functions in eukaryotic cells. Herein, we focus on the role of non-receptor protein tyrosine kinases, most notably, FAK, c-Yes and c-Src, in the transport of spermatids across the seminiferous epithelium during spermatogenesis. Since spermatids, which are formed from spermatocytes via meiosis, are immotile haploid cells, they must be transported by Sertoli cells across the seminiferous epithelium during the epithelial cycle of spermatogenesis. Without the timely transport of spermatids across the epithelium, the release of sperms at spermiation fails to occur, leading to infertility. Thus, the molecular event pertinent to spermatid transport is crucial to spermatogenesis. We provide a critical discussion based on recent findings in this review. We also provide a hypothetical model on spermatid transport, and the role of non-receptor protein tyrosine kinases in this event. We also highlight areas of research that deserve attention by investigators in the field.


Assuntos
Proteínas Tirosina Quinases/fisiologia , Transporte Espermático , Espermátides/enzimologia , Espermatogênese , Animais , Barreira Hematotesticular/citologia , Barreira Hematotesticular/fisiologia , Humanos , Masculino , Fosforilação , Processamento de Proteína Pós-Traducional , Epitélio Seminífero/citologia , Células de Sertoli/enzimologia , Transdução de Sinais , Espermátides/fisiologia
4.
Am J Physiol Endocrinol Metab ; 307(7): E553-62, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25117412

RESUMO

The blood-testis barrier (BTB) is one of the tightest blood-tissue barriers in the mammalian body. However, it undergoes cyclic restructuring during the epithelial cycle of spermatogenesis in which the "old" BTB located above the preleptotene spermatocytes being transported across the immunological barrier is "disassembled," whereas the "new" BTB found behind these germ cells is rapidly "reassembled," i.e., mediated by endocytic vesicle-mediated protein trafficking events. Thus, the immunological barrier is maintained when preleptotene spermatocytes connected in clones via intercellular bridges are transported across the BTB. Yet the underlying mechanism(s) in particular the involving regulatory molecules that coordinate these events remains unknown. We hypothesized that c-Src and c-Yes might work in contrasting roles in endocytic vesicle-mediated trafficking, serving as molecular switches, to effectively disassemble and reassemble the old and the new BTB, respectively, to facilitate preleptotene spermatocyte transport across the BTB. Following siRNA-mediated specific knockdown of c-Src or c-Yes in Sertoli cells, we utilized biochemical assays to assess the changes in protein endocytosis, recycling, degradation and phagocytosis. c-Yes was found to promote endocytosed integral membrane BTB proteins to the pathway of transcytosis and recycling so that internalized proteins could be effectively used to assemble new BTB from the disassembling old BTB, whereas c-Src promotes endocytosed Sertoli cell BTB proteins to endosome-mediated protein degradation for the degeneration of the old BTB. By using fluorescence beads mimicking apoptotic germ cells, Sertoli cells were found to engulf beads via c-Src-mediated phagocytosis. A hypothetical model that serves as the framework for future investigation is thus proposed.


Assuntos
Barreira Hematotesticular/metabolismo , Proteínas Proto-Oncogênicas c-yes/fisiologia , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Células de Sertoli/metabolismo , Vesículas Transportadoras/metabolismo , Animais , Técnicas de Cultura de Células , Células Cultivadas , Endocitose/fisiologia , Técnicas de Silenciamento de Genes , Genes src/genética , Masculino , Proteínas de Membrana/metabolismo , Fagocitose/fisiologia , Proteínas Proto-Oncogênicas c-yes/genética , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley
5.
FASEB J ; 27(2): 464-77, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23073828

RESUMO

Development of spermatozoa in adult mammalian testis during spermatogenesis involves extensive cell migration and differentiation. Spermatogonia that reside at the basal compartment of the seminiferous epithelium differentiate into more advanced germ cell types that migrate toward the apical compartment until elongated spermatids are released into the tubule lumen during spermiation. Apical ectoplasmic specialization (ES; a testis-specific anchoring junction) is the only cell junction that anchors and maintains the polarity of elongating/elongated spermatids (step 8-19 spermatids) in the epithelium. Little is known regarding the signaling pathways that trigger the disassembly of the apical ES at spermiation. Here, we show that secreted Frizzled-related protein 1 (sFRP1), a putative tumor suppressor gene that is frequently down-regulated in multiple carcinomas, is a crucial regulatory protein for spermiation. The expression of sFRP1 is tightly regulated in adult rat testis to control spermatid adhesion and sperm release at spermiation. Down-regulation of sFRP1 during testicular development was found to coincide with the onset of the first wave of spermiation at approximately age 45 d postpartum, implying that sFRP1 might be correlated with elongated spermatid adhesion conferred by the apical ES before spermiation. Indeed, administration of sFRP1 recombinant protein to the testis in vivo delayed spermiation, which was accompanied by down-regulation of phosphorylated (p)-focal adhesion kinase (FAK)-Tyr(397) and retention of nectin-3 adhesion protein at the apical ES. To further investigate the functional relationship between p-FAK-Tyr(397) and localization of nectin-3, we overexpressed sFRP1 using lentiviral vectors in the Sertoli-germ cell coculture system. Consistent with the in vivo findings, overexpression of sFRP1 induced down-regulation of p-FAK-Tyr(397), leading to a decline in phosphorylation of nectin-3. In summary, this report highlights the critical role of sFRP1 in regulating spermiation via its effects on the FAK signaling and retention of nectin-3 adhesion complex at the apical ES.


Assuntos
Moléculas de Adesão Celular/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Espermátides/metabolismo , Testículo/metabolismo , Animais , Sequência de Bases , Adesão Celular/fisiologia , Técnicas de Cocultura , DNA Complementar/genética , Quinase 1 de Adesão Focal/química , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Proteínas de Membrana/genética , Microscopia Eletrônica de Transmissão , Complexos Multiproteicos/metabolismo , Nectinas , Fosforilação , Ratos , Ratos Sprague-Dawley , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Transdução de Sinais , Espermátides/citologia , Espermatogênese/fisiologia , Testículo/citologia , Testículo/crescimento & desenvolvimento
6.
bioRxiv ; 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38895201

RESUMO

Transposable elements (TEs) are abundant in the human genome, and they provide the sources for genetic and functional diversity. The regulation of TEs expression and their functional consequences in physiological conditions and cancer development remain to be fully elucidated. Previous studies suggested TEs are repressed by DNA methylation and chromatin modifications. The effect of 3D chromatin topology on TE regulation remains elusive. Here, by integrating transcriptome and 3D genome architecture studies, we showed that haploinsufficient loss of NIPBL selectively activates alternative promoters at the long terminal repeats (LTRs) of the TE subclasses. This activation occurs through the reorganization of topologically associating domain (TAD) hierarchical structures and recruitment of proximal enhancers. These observations indicate that TAD hierarchy restricts transcriptional activation of LTRs that already possess open chromatin features. In cancer, perturbation of the hierarchical chromatin topology can lead to co-option of LTRs as functional alternative promoters in a context-dependent manner and drive aberrant transcriptional activation of novel oncogenes and other divergent transcripts. These data uncovered a new layer of regulatory mechanism of TE expression beyond DNA and chromatin modification in human genome. They also posit the TAD hierarchy dysregulation as a novel mechanism for alternative promoter-mediated oncogene activation and transcriptional diversity in cancer, which may be exploited therapeutically.

7.
Am J Physiol Endocrinol Metab ; 304(7): E757-69, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23403943

RESUMO

Drug transporters determine the bioavailability of drugs in the testis behind the blood-testis barrier (BTB). Thus, they are crucial for male contraceptive development if these drugs (e.g., adjudin) exert their effects behind the BTB. Herein breast cancer resistance protein (Bcrp), an efflux drug transporter, was found to be expressed by both Sertoli and germ cells. Interestingly, Bcrp was not a component of the Sertoli cell BTB. Instead, it was highly expressed by peritubular myoid cells at the tunica propria and also endothelial cells of the microvessels in the interstitium at all stages of the epithelial cycle. Unexpectedly, Bcrp was found to be expressed at the Sertoli-step 18-19 spermatid interface but limited to stage VI-early VIII tubules, and an integrated component of the apical ectoplasmic specialization (apical ES). Apparently, Bcrp is being used by late-stage spermatids to safeguard their completion of spermiogenesis by preventing harmful drugs to enter these cells while they transform to spermatozoa. Also, the association of Bcrp with actin, Eps8 (epidermal growth factor receptor pathway substrate 8, an actin barbed end capping and bundling protein), and Arp3 (actin-related protein 3, a component of the Arp2/3 complex known to induce branched actin polymerization) at the apical ES suggest that Bcrp may be involved in regulating the organization of actin filament bundles at the site. Indeed, a knockdown of Bcrp by RNAi in the testis perturbed the apical ES function, disrupting spermatid polarity and adhesion. In summary, Bcrp is a regulator of the F-actin-rich apical ES in the testis.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Barreira Hematotesticular/metabolismo , Células Endoteliais/metabolismo , Epitélio Seminífero/metabolismo , Células de Sertoli/metabolismo , Espermatogênese/fisiologia , Espermatozoides/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/efeitos dos fármacos , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteína 3 Relacionada a Actina/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Anticoncepcionais Masculinos/farmacologia , Células Endoteliais/citologia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Hidrazinas/farmacologia , Indazóis/farmacologia , Masculino , Microvasos/citologia , Microvasos/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Epitélio Seminífero/citologia , Células de Sertoli/citologia , Espermátides/citologia , Espermátides/metabolismo , Espermatogênese/genética , Espermatozoides/citologia
8.
Proc Natl Acad Sci U S A ; 107(25): 11399-404, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20534521

RESUMO

In the testis, the blood-testis barrier (BTB) is constituted by specialized junctions between adjacent Sertoli cells in the seminiferous epithelium near the basement membrane. Although the BTB is one of the tightest blood-tissue barriers in the mammalian body, it undergoes extensive restructuring at stage VIII of the seminiferous epithelial cycle to facilitate the transit of preleptotene spermatocytes. Thus, meiosis and postmeiotic germ cell development take place in the seminiferous epithelium behind the BTB. Cytokines (e.g., TGF-beta3) are known to regulate BTB dynamics by enhancing the endocytosis of integral membrane proteins and their intracellular degradation. This thus reduces the levels of proteins above the spermatocytes in transit at the BTB, causing its disruption after testosterone-induced new tight junction (TJ) fibrils are formed behind these cells. By using Sertoli cells cultured in vitro with an established TJ permeability barrier that mimicked the BTB in vivo, Cdc42 was shown to be a crucial regulator that mediated the TGF-beta3-induced BTB disruption. TGF-beta3 was shown to activate Cdc42 to its active GTP-bound form. However, an inactivation of Cdc42 by overexpressing its dominant-negative mutant T17N in Sertoli cell epithelium was shown to block the TGF-beta3-induced acceleration in protein endocytosis. Consequently, this prevented the disruption of Sertoli cell TJ permeability barrier and redistribution of TJ proteins (e.g., CAR and ZO-1) from the cell-cell interface to cell cytosol caused by TGF-beta3. In summary, Cdc42 is a crucial regulatory component in the TGF-beta3-mediated cascade of events that leads to the disruption of the TJ fibrils above the preleptotene spermatocytes to facilitate their transit.


Assuntos
Barreira Hematotesticular/metabolismo , Sangue/metabolismo , Testículo/metabolismo , Fator de Crescimento Transformador beta3/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Endocitose , Células Epiteliais/metabolismo , Guanosina Trifosfato/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Epitélio Seminífero/metabolismo , Células de Sertoli/metabolismo , Espermatócitos/metabolismo , Junções Íntimas/metabolismo
9.
Sci Adv ; 9(14): eadc9446, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-37018402

RESUMO

The mechanisms underlying ETS-driven prostate cancer initiation and progression remain poorly understood due to a lack of model systems that recapitulate this phenotype. We generated a genetically engineered mouse with prostate-specific expression of the ETS factor, ETV4, at lower and higher protein dosage through mutation of its degron. Lower-level expression of ETV4 caused mild luminal cell expansion without histologic abnormalities, and higher-level expression of stabilized ETV4 caused prostatic intraepithelial neoplasia (mPIN) with 100% penetrance within 1 week. Tumor progression was limited by p53-mediated senescence and Trp53 deletion cooperated with stabilized ETV4. The neoplastic cells expressed differentiation markers such as Nkx3.1 recapitulating luminal gene expression features of untreated human prostate cancer. Single-cell and bulk RNA sequencing showed that stabilized ETV4 induced a previously unidentified luminal-derived expression cluster with signatures of cell cycle, senescence, and epithelial-to-mesenchymal transition. These data suggest that ETS overexpression alone, at sufficient dosage, can initiate prostate neoplasia.


Assuntos
Neoplasia Prostática Intraepitelial , Neoplasias da Próstata , Masculino , Camundongos , Animais , Humanos , Próstata/metabolismo , Próstata/patologia , Proteína Supressora de Tumor p53/metabolismo , Neoplasias da Próstata/genética , Fatores de Transcrição/metabolismo , Neoplasia Prostática Intraepitelial/genética , Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Proto-Oncogênicas c-ets/genética
10.
Adv Exp Med Biol ; 763: 149-70, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23397623

RESUMO

The Scribble polarity complex or module is one of the three polarity modules that regulate cell polarity in multiple epithelia including blood-tissue barriers. This protein complex is composed of Scribble, Lethal giant larvae (Lgl) and Discs large (Dlg), which are well conserved across species from fruitflies and worms to mammals. Originally identified in Drosophila and C. elegans where the Scribble complex was found to work with the Par-based and Crumbs-based polarity modules to regulate apicobasal polarity and asymmetry in cells and tissues during embryogenesis, their mammalian homologs have all been identified in recent years. Components of the Scribble complex are known to regulate multiple cellular functions besides cell polarity, which include cell proliferation, assembly and maintenance of adherens junction (AJ) and tight junction (TJ), and they are also tumor suppressors. Herein, we provide an update on the Scribble polarity complex and how this protein complex modulates cell adhesion with some emphasis on its role in Sertoli cell blood-testis barrier (BTB) function. It should be noted that this is a rapidly developing field, in particular the role of this protein module in blood-tissue barriers, and this short chapter attempts to provide the information necessary for investigators studying reproductive biology and blood-tissue barriers to design future studies. We also include results of recent studies from flies and worms since this information will be helpful in planning experiments for future functional studies in the testis to understand how Scribble-based proteins regulate BTB dynamics and spermatogenesis.


Assuntos
Barreira Hematotesticular/metabolismo , Polaridade Celular , Proteínas de Drosophila/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de Membrana/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Barreira Hematotesticular/citologia , Adesão Celular , Proliferação de Células , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imuno-Histoquímica , Mucosa Intestinal/citologia , Mamíferos/metabolismo , Proteínas de Membrana/genética , Complexos Multiproteicos/genética , Mutação , Estrutura Terciária de Proteína , Transporte Proteico , Transdução de Sinais , Espermatogênese , Junções Íntimas/genética , Junções Íntimas/metabolismo , Proteínas Supressoras de Tumor/genética
11.
Proc Natl Acad Sci U S A ; 106(23): 9298-303, 2009 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-19470647

RESUMO

In mammalian testes, such as rats, the mechanism(s) that regulate blood-testis barrier (BTB) restructuring at stages VIII-IX of the seminiferous epithelial cycle of spermatogenesis to facilitate the transit of preleptotene/leptotene spermatocytes is not known. This is due to the lack of information on the regulatory proteins at the BTB. Herein, focal adhesion kinase (FAK), a nonreceptor protein tyrosine kinase, is shown to structurally interact with occludin and ZO-1 to form a functional protein complex at the BTB. Its expression at the BTB in the seminiferous epithelium is stage specific, being lowest at stage VIII-IX tubules, analogous to the expression pattern of occludin. Using primary Sertoli cells cultured in vitro with an established tight junction (TJ) permeability barrier that mimics the BTB in vivo, the knockdown of FAK by RNAi led to a transient disruption of the TJ barrier. This was accompanied by a loss of association between occludin and ZO-1, likely the result of reduced occludin phosphorylation at Tyr and Ser residues, but not Thr, which in turn led to a redistribution of occludin at the Sertoli-Sertoli cell interface, moving from cell membrane into cell cytosol, thereby disrupting the BTB. These findings suggest that a similar mechanism is in place in the testis in vivo to regulate BTB restructuring to facilitate the transit of primary spermatocytes. Furthermore, FAK was shown to be a molecular target of cadmium because its knockdown would desensitize Sertoli cells to cadmium-induced TJ barrier disruption. In summary, FAK is a unique regulator of BTB dynamics in the testis.


Assuntos
Barreira Hematotesticular/fisiologia , Animais , Técnicas de Silenciamento de Genes , Masculino , Proteínas de Membrana/metabolismo , Ocludina , Fosfoproteínas/metabolismo , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Células de Sertoli/metabolismo , Espermatogênese , Testículo/citologia , Testículo/fisiologia , Junções Íntimas/metabolismo , Proteína da Zônula de Oclusão-1
12.
Cancer Discov ; 12(9): 2120-2139, 2022 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-35789380

RESUMO

Polycomb repressive complex 2 (PRC2) has oncogenic and tumor-suppressive roles in cancer. There is clinical success of targeting this complex in PRC2-dependent cancers, but an unmet therapeutic need exists in PRC2-loss cancer. PRC2-inactivating mutations are a hallmark feature of high-grade malignant peripheral nerve sheath tumor (MPNST), an aggressive sarcoma with poor prognosis and no effective targeted therapy. Through RNAi screening in MPNST, we found that PRC2 inactivation increases sensitivity to genetic or small-molecule inhibition of DNA methyltransferase 1 (DNMT1), which results in enhanced cytotoxicity and antitumor response. Mechanistically, PRC2 inactivation amplifies DNMT inhibitor-mediated expression of retrotransposons, subsequent viral mimicry response, and robust cell death in part through a protein kinase R (PKR)-dependent double-stranded RNA sensor. Collectively, our observations posit DNA methylation as a safeguard against antitumorigenic cell-fate decisions in PRC2-loss cancer to promote cancer pathogenesis, which can be therapeutically exploited by DNMT1-targeted therapy. SIGNIFICANCE: PRC2 inactivation drives oncogenesis in various cancers, but therapeutically targeting PRC2 loss has remained challenging. Here we show that PRC2-inactivating mutations set up a tumor context-specific liability for therapeutic intervention via DNMT1 inhibitors, which leads to innate immune signaling mediated by sensing of derepressed retrotransposons and accompanied by enhanced cytotoxicity. See related commentary by Guil and Esteller, p. 2020. This article is highlighted in the In This Issue feature, p. 2007.


Assuntos
Antineoplásicos , Neoplasias , Neurofibrossarcoma , Carcinogênese/genética , Humanos , Mutação , Neoplasias/genética , Neurofibrossarcoma/diagnóstico , Neurofibrossarcoma/genética , Neurofibrossarcoma/patologia , Complexo Repressor Polycomb 2/genética , Retroelementos
13.
Proc Natl Acad Sci U S A ; 105(28): 9657-62, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18621709

RESUMO

The Par3/Par6/aPKC and the CRB3/Pals1/PATJ polarity complexes are involved in regulating apical ectoplasmic specialization (ES) and blood-testis barrier (BTB) restructuring in the testis. Par6 was a component of the apical ES and the BTB. However, its level was considerably diminished at both sites at stage VIII of the cycle. Par6 also formed a stable complex with Pals1 and JAM-C (a component of the apical ES) in normal testes. When rats were treated with adjudin to induce apical ES restructuring without compromising the BTB, Par6 staining virtually disappeared at the apical ES in misaligned spermatids before their depletion. Additionally, the Par6/Pals1 complex became tightly associated with Src kinase, rendering a loss of association of the Par6/Pals1 complex with JAM-C, thereby destabilizing apical ES to facilitate spermatid loss. Primary Sertoli cell cultures with established functional BTB, but without apical ES, were next used to assess the Par6-based complex on BTB dynamics. When either Par6 or Par3 was knocked down by RNAi in Sertoli cell epithelium, a significant loss of the corresponding protein by approximately 60% in cells vs. controls was detected, alongside with a decline in aPKC after Par6, but not Par3, knockdown. This Par3 or Par6 knockdown also led to a transient loss of selected BTB proteins at the cell-cell interface, thereby compromising the BTB integrity. These findings illustrate that the Par6/Par3-based polarity complex likely coordinates the events of apical ES and BTB restructuring that take place concurrently at the opposing ends of adjacent Sertoli cells in the seminiferous epithelium during spermatogenesis.


Assuntos
Barreira Hematotesticular/metabolismo , Proteínas de Transporte/fisiologia , Polaridade Celular , Espermatogênese , Proteínas Adaptadoras de Transdução de Sinal , Animais , Movimento Celular , Hidrazinas/farmacologia , Indazóis/farmacologia , Masculino , Proteínas do Tecido Nervoso , Ratos , Espermátides/citologia , Testículo/citologia
14.
Proc Natl Acad Sci U S A ; 105(26): 8950-5, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18579774

RESUMO

The mechanism(s) that regulate and coordinate the events of spermiation and blood-testis barrier (BTB) restructuring in the seminiferous epithelium that occur concurrently at stage VIII of the seminiferous epithelial cycle of spermatogenesis are unknown. In this report, fragments derived from the laminin complex composed of laminin alpha3, beta3, and gamma3 chains (laminin-333) at the apical ectoplasmic specialization (apical ES) were shown to modulate BTB dynamics directly and/or indirectly via hemidesmosome. Experiments were performed using cultured Sertoli cells with functional tight junction (TJ) barrier and the ultrastructural features of the BTB but not apical ES. Recombinant protein fragments of laminin beta3 and gamma3 chains were shown to reduce the protein levels of occludin and beta1-integrin dose dependently at the Sertoli-Sertoli and Sertoli-basement membrane interface, respectively, thereby destabilizing the BTB permeability function. These results were corroborated by transient overexpression of laminin fragments in Sertoli cells. To further assess the role of beta1-integrin in hemidesmosome, knockdown of beta1-integrin in Sertoli cells by RNAi was found to associate with occludin redistribution at the Sertoli-Sertoli cell interface, wherein occludin moved away from the cell surface and became associated with endosomes, thereby destabilizing the BTB. In short, an apical ES-BTB-hemidesmosome autocrine regulatory axis was identified in testes, coordinating the events of spermiation and BTB restructuring that occur at the opposite ends of the seminiferous epithelium during spermatogenesis.


Assuntos
Comunicação Autócrina , Barreira Hematotesticular/metabolismo , Espermatogênese , Animais , Células Cultivadas , Integrina beta1/metabolismo , Laminina/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Modelos Biológicos , Ocludina , Fragmentos de Peptídeos/metabolismo , Transporte Proteico , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Epitélio Seminífero/citologia , Epitélio Seminífero/metabolismo , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Junções Íntimas/metabolismo
15.
Dev Biol ; 327(1): 48-61, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19103189

RESUMO

In adult mammals such as rats, the blood-testis barrier (BTB) conferred by adjacent Sertoli cells in the seminiferous epithelium segregates post-meiotic germ cell development from the systemic circulation and is one of the tightest blood-tissue barriers. Yet it must "open" transiently at stages VIII to IX of the epithelial cycle to accommodate the migration of preleptotene/leptotene spermatocytes. While this is a vital event of spermatogenesis, the mechanism(s) that regulates BTB dynamics is virtually unknown. Recent studies have suggested that transforming growth factor-beta3 (TGF-beta3) and tumor necrosis factor alpha (TNFalpha) secreted by Sertoli and germ cells into the microenvironment of the BTB are capable of inducing reversible BTB disruption in vivo, apparently by reducing the steady-state levels of occludin and zonula occludens-1 (ZO-1) at the BTB via the p38 mitogen activated protein (MAP) kinase signaling pathway. In this study, local administration of TGF-beta3 (200 ng/testis) to the testis was shown to reversibly perturb the BTB integrity in vivo. We next sought to delineate the mechanism by which these cytokines maintain the steady-state level of integral membrane proteins: occludin, junctional adhesion molecule-A (JAM-A) and N-cadherin at the BTB. Primary Sertoli cells cultured in vitro were shown to establish intact tight junctions and functional BTB within two days when assessed by transepithelial electrical resistance (TER) measurement across the cell epithelium. Sertoli cell integral membrane protein internalization at the BTB was assessed by biotinylation of cell surface proteins, to be followed by tracking the endocytosed/biotinylated proteins by using specific antibodies. Both TGF-beta3 (3 ng/ml) and TNFalpha (10 ng/ml) were shown to significantly accelerate the kinetics of internalization of JAM-A, N-cadherin, and occludin versus controls. Treatment of cells with phenylarsine oxide (PAO) at 10 microM that blocks clathrin-mediated endocytosis was shown to inhibit the TGF-beta3-induced protein internalization. This inhibition of TGF-beta3-mediated protein endocytosis was further validated by silencing of clathrin. The specific effect of TGF-beta3 on protein internalization was further confirmed by RNAi using specific TGF-beta receptor I (TbetaR1) siRNA duplexes. When TbetaR1 was knocked down, the TGF-beta3-induced increase in the kinetics of JAM-A and occludin endocytosis was abolished, making them indistinguishable from controls, illustrating the specificity of the TGF-beta3 effects on protein endocytosis. In summary, this report demonstrates for the first time that BTB dynamics are regulated by TGF-beta3 and TNFalpha via an enhancement of protein endocytosis at the BTB.


Assuntos
Barreira Hematotesticular/efeitos dos fármacos , Fator de Crescimento Transformador beta3/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Clatrina , Endocitose/efeitos dos fármacos , Masculino , Proteínas de Membrana/metabolismo , Ratos , Ratos Sprague-Dawley , Espermatogênese , Testículo
16.
Biochim Biophys Acta ; 1778(3): 692-708, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18068662

RESUMO

Anchoring junctions are cell adhesion apparatus present in all epithelia and endothelia. They are found at the cell-cell interface (adherens junction (AJ) and desmosome) and cell-matrix interface (focal contact and hemidesmosome). In this review, we focus our discussion on AJ in particular the dynamic changes and regulation of this junction type in normal epithelia using testis as a model. There are extensive restructuring of AJ (e.g., ectoplasmic specialization, ES, a testis-specific AJ) at the Sertoli-Sertoli cell interface (basal ES) and Sertoli-elongating spermatid interface (apical ES) during the seminiferous epithelial cycle of spermatogenesis to facilitate the migration of developing germ cells across the seminiferous epithelium. Furthermore, recent findings have shown that ES also confers cell orientation and polarity in the seminiferous epithelium, illustrating that some of the functions initially ascribed to tight junctions (TJ), such as conferring cell polarity, are also part of the inherent properties of the AJ (e.g., apical ES) in the testis. The biology and regulation based on recent studies in the testis are of interest to cell biologists in the field, in particular their regulation, which perhaps is applicable to tumorigenesis.


Assuntos
Junções Aderentes/fisiologia , Junções Aderentes/ultraestrutura , Testículo/fisiologia , Testículo/ultraestrutura , Animais , Polaridade Celular , Endocitose , Homeostase , Humanos , Masculino , Proteínas de Membrana/química , Proteínas de Membrana/fisiologia , Modelos Biológicos , Complexos Multiproteicos , Peptídeo Hidrolases/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Inibidores de Proteases/metabolismo , Proteínas Quinases/fisiologia , Epitélio Seminífero/fisiologia , Epitélio Seminífero/ultraestrutura , Espermatogênese
17.
Curr Med Chem ; 16(7): 906-15, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19275601

RESUMO

Even though various contraceptive methods are widely available, the number of unwanted pregnancies is still on the rise in developing countries, pressurizing the already resource limited nations. One of the major underlying reasons is the lack of effective, low cost, and safe contraceptives for couples. During the past decade, some studies were performed using animal models to decipher if the Sertoli-germ cell junction in the testis is a target for male fertility regulation. Some of these study models were based on the use of hormones and/or chemicals to disrupt the hypothalamic-pituitary-testicular axis (e.g., androgen-based implants or pills) and others utilized a panel of chemical entities or synthetic peptides to perturb spermatogenesis either reversibly or non-reversibly. Among them, adjudin, a potential male contraceptive, is one of the compounds exerting its action on the unique adherens junctions, known as ectoplasmic specializations, in the testis. Since the testis is equipped with inter-connected cell junctions, an initial targeting of one junction type may affect the others and these accumulative effects could lead to spermatogenic arrest. This review attempts to cover an innovative theme on how male infertility can be achieved by inducing junction instability and defects in the testis, opening a new window of research for male contraceptive development. While it will still take much time and effort of intensive investigation before a product can reach the consumable market, these findings have provided hope for better family planning involving men.


Assuntos
Anticoncepcionais Masculinos/farmacologia , Testículo/efeitos dos fármacos , Testículo/fisiologia , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/fisiologia , Animais , Humanos , Masculino , Modelos Animais , Ratos , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/fisiologia , Espermatogênese/efeitos dos fármacos , Espermatogênese/fisiologia
18.
Cell Syst ; 8(5): 446-455.e8, 2019 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-31078526

RESUMO

Recent studies have shown that mutations at non-coding elements, such as promoters and enhancers, can act as cancer drivers. However, an important class of non-coding elements, namely CTCF insulators, has been overlooked in the previous driver analyses. We used insulator annotations from CTCF and cohesin ChIA-PET and analyzed somatic mutations in 1,962 whole genomes from 21 cancer types. Using the heterogeneous patterns of transcription-factor-motif disruption, functional impact, and recurrence of mutations, we developed a computational method that revealed 21 insulators showing signals of positive selection. In particular, mutations in an insulator in multiple cancer types, including 16% of melanoma samples, are associated with TGFB1 up-regulation. Using CRISPR-Cas9, we find that alterations at two of the most frequently mutated regions in this insulator increase cell growth by 40%-50%, supporting the role of this boundary element as a cancer driver. Thus, our study reveals several CTCF insulators as putative cancer drivers.


Assuntos
Fator de Ligação a CCCTC/genética , Fator de Ligação a CCCTC/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Genoma Humano , Humanos , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Repressoras/genética , Coesinas
19.
Cancer Cell ; 35(4): 603-617.e8, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30930119

RESUMO

Deletion of the gene encoding the chromatin remodeler CHD1 is among the most common alterations in prostate cancer (PCa); however, the tumor-suppressive functions of CHD1 and reasons for its tissue-specific loss remain undefined. We demonstrated that CHD1 occupied prostate-specific enhancers enriched for the androgen receptor (AR) and lineage-specific cofactors. Upon CHD1 loss, the AR cistrome was redistributed in patterns consistent with the oncogenic AR cistrome in PCa samples and drove tumor formation in the murine prostate. Notably, this cistrome shift was associated with a unique AR transcriptional signature enriched for pro-oncogenic pathways unique to this tumor subclass. Collectively, these data credential CHD1 as a tumor suppressor in the prostate that constrains AR binding/function to limit tumor progression.


Assuntos
Carcinogênese , DNA Helicases/deficiência , Proteínas de Ligação a DNA/deficiência , Elementos Facilitadores Genéticos , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Transcrição Gênica , Proteínas Supressoras de Tumor/deficiência , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Ligação Proteica , Receptores Androgênicos/genética , Transdução de Sinais , Técnicas de Cultura de Tecidos , Proteínas Supressoras de Tumor/genética
20.
Cancer Discov ; 8(2): 234-251, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29162563

RESUMO

The cellular context that integrates upstream signaling and downstream nuclear response dictates the oncogenic behavior and shapes treatment responses in distinct cancer types. Here, we uncover that in gastrointestinal stromal tumor (GIST), the forkhead family member FOXF1 directly controls the transcription of two master regulators, KIT and ETV1, both required for GIST precursor-interstitial cells of Cajal lineage specification and GIST tumorigenesis. Further, FOXF1 colocalizes with ETV1 at enhancers and functions as a pioneer factor that regulates the ETV1-dependent GIST lineage-specific transcriptome through modulation of the local chromatin context, including chromatin accessibility, enhancer maintenance, and ETV1 binding. Functionally, FOXF1 is required for human GIST cell growth in vitro and murine GIST tumor growth and maintenance in vivo The simultaneous control of the upstream signaling and nuclear response sets up a unique regulatory paradigm and highlights the critical role of FOXF1 in enforcing the GIST cellular context for highly lineage-restricted clinical behavior and treatment response.Significance: We uncover that FOXF1 defines the core-regulatory circuitry in GIST through both direct transcriptional regulation and pioneer factor function. The unique and simultaneous control of signaling and transcriptional circuitry by FOXF1 sets up an enforced transcriptional addiction to FOXF1 in GIST, which can be exploited diagnostically and therapeutically. Cancer Discov; 8(2); 234-51. ©2017 AACR.See related commentary by Lee and Duensing, p. 146This article is highlighted in the In This Issue feature, p. 127.


Assuntos
Fatores de Transcrição Forkhead/genética , Tumores do Estroma Gastrointestinal/genética , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Animais , Biomarcadores Tumorais , Ciclo Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Tumores do Estroma Gastrointestinal/metabolismo , Perfilação da Expressão Gênica , Xenoenxertos , Humanos , Ligação Proteica , Transdução de Sinais , Fatores de Transcrição/genética , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa